May 15 marks International Huntington's Disease Awareness Day — a date not only meant to raise public awareness for those affected but also to highlight the urgent need for breakthroughs in treating this devastating condition. Known as the “disease of dancing,” Huntington's causes patients to gradually lose control over their bodies through involuntary movements, as if pulled by invisible strings, step by step into the decline of motor skills and cognitive functions. Globally, 3 to 7 people per 100,000 are enduring this suffering, while effective medical solutions remain elusive.
Huntingtin (HTT) is a disease-associated gene linked to Huntington's disease (HD). It is widely expressed across various tissues and organs throughout the body, including the central nervous system, and is essential for normal development. Near the 5’end of the HTT gene's coding region lies a polymorphic trinucleotide repeat sequence—cytosine-adenine-guanine (CAG)—which, during translation, encodes a polyglutamine (polyQ) tract.
Huntington's disease is a neurodegenerative disorder characterized by the loss of neurons in the striatum. It is caused by abnormal expression of the trinucleotide (CAG) repeat sequence in the HTT gene. When the number of CAG repeats exceeds 35, it leads to an abnormal expansion of the polyglutamine (polyQ) tract, resulting in the misfolding of HTT protein fragments. These misfolded proteins abnormally interact with numerous other proteins and accumulate in the cell nucleus and neuronal terminals, disrupting neurotransmission, intracellular protein transport, and mitochondrial function. Ultimately, this cascade leads to neuronal dysfunction and degeneration.
In recent years, drug development for Huntington’s disease has focused on eliminating or functionally correcting the mutant huntingtin protein (mHTT). The main therapeutic strategies include gene-silencing approaches—such as Roche’s antisense oligonucleotide (ASO) drug Tominersen and Wave Life Sciences’RNAi therapy WVE-003—as well as small-molecule drugs like PTC518, which promotes mHTT degradation, and ANX005, which targets neuroinflammation. Cutting-edge efforts also explore gene editing technologies and stem cell transplantation.
Although Tominersen did not meet expectations in its Phase III clinical trial, it provided critical data for target validation. Current research efforts are shifting toward optimizing delivery systems—such as AAV vectors capable of crossing the blood-brain barrier—and exploring combination therapies that synergize protein clearance with anti-inflammatory mechanisms. With the application of novel technologies like base editing and the advancement of adaptive clinical trials, the field is steadily overcoming key challenges and moving toward more precise and targeted therapies for Huntington's disease.
However, optimizing and advancing these therapeutic strategies requires a deeper understanding of the pathological mechanisms of Huntington’s disease, along with the development of experimental models that accurately recapitulate the disease's characteristics.
To support this need, we have developed the hHTT Q150 knock-in model—FVB-hHTT Q150 KI mice (Product No.: I001019). Using gene editing technology, a mutant human HTT gene sequence carrying 150 CAG repeats is precisely knocked into the mouse genome.
According to published studies, this mouse model exhibits pathological features and functional impairments characteristic of Huntington’s disease, making it a valuable tool for drug discovery, efficacy screening, and safety evaluation in HD research.
Figure 1. Locomotor activity (A–C) and percentage of time spent in the central area (D–F) during the open field test in wild-type (WT) mice and FVB-hHTT Q150 KI mice.
Figure 2. Central area movement speed (A–C) and peripheral area movement speed (D–F) in the open field test for wild-type (WT) mice and FVB-hHTT Q150 KI mice.
Figure 3. Total distance traveled (A–C) and percentage of time spent in the central area (D–F) during the open field test in wild-type (WT) mice and FVB-hHTT Q150 KI mice.
Figure 4. Central area movement speed (A–C) and peripheral area movement speed (D–F) during the open field test in wild-type (WT) mice and FVB-hHTT Q150 KI mice.
Figure 5. Latency to fall in the rotarod test for wild-type (WT) mice and FVB-hHTT Q150 KI mice.
Figure 6. Grip strength, rotarod performance, and open field analysis of 14-month-old hHTT-Q150 KI mice.
Leveraging our well-established animal model development platforms, we offer over 2,000 ready-to-use KO/CKO mouse models related to the nervous system, as well as more than 20 types of genetically engineered and drug-induced rodent models for neurological diseases. These models encompass a wide range of targeting strategies, including knockout, conditional knockout, point mutation, transgenic, and humanized models. In addition to our off-the-shelf offerings, we also provide customized model generation and collaborative development services tailored to specific research needs.
Product Number | Product | Strain Background | Application |
C001427 | B6-hSNCA | C57BL/6NCya | Parkinson's disease |
C001504 | B6-hSMN2(SMA) | C57BL/6NCya | Spinal muscular atrophy (SMA) |
C001518 | DMD-Q995* | C57BL/6JCya | Duchenne muscular dystrophy (DMD) |
C001410 | B6-htau | C57BL/6JCya | Frontotemporal dementia, Alzheimer's disease, and other neurodegenerative diseases |
C001437 | B6-hIGHMBP2 | C57BL/6NCya | Spinal muscular atrophy with respiratory distress type 1 and Charcot-Marie-Tooth disease type 2S |
C001418 | B6-hTARDBP | C57BL/6JCya | Amyotrophic lateral sclerosis, frontotemporal dementia, and other neurodegenerative diseases |
C001398 | B6-hATXN3 | C57BL/6NCya | Spinocerebellar ataxia type 3 |
C001568 | B6-hMECP2 | C57BL/6NCya | Rett syndrome |
C001569 | B6-hMECP2*T158M | C57BL/6NCya | Rett syndrome |
I001124 | B6-hLMNA | C57BL/6NCya | Progeria syndrome |
CG0015 | 6-OHDA Treated Rats | - | Parkinson's disease (PD) |
CG0016 | CUMS Model | C57BL/6JCya | Depression |
C001210 | AD-M1 | C57BL/6JCya | Research on Alzheimer's Disease (AD), Cerebral Amyloid Angiopathy (CAA) and Notch signaling pathway. |
C001541 | AD-M2 | C57BL/6JCya | Research on Alzheimer's Disease (AD), Cerebral Amyloid Angiopathy (CAA), Notch signaling pathway and other neurodegenerative diseases. |
I001019 | FVB-hHTT Q150 KI | FVB/NJCya | Development and screening of therapeutic drugs for Huntington's disease; Evaluation of therapeutic drug efficacy and safety for Huntington's disease; Research on the pathogenesis of Huntington's disease. |
- | MPTP-treated Mice | - | Parkinson's disease (PD) |
- | Chronic Compression Injury Model of the Sciatic Nerve (CCI) | - | - |
C001582 | Mecp2 KO | C57BL/6JCya | Rett syndrome (RTT) |
C001611 | Ube3a KO | C57BL/6NCya |
|
C001397 | TG-hATXN3(85Q) | C57BL/6JCya | Spinocerebellar ataxia type 3,SCA3 |